1
|
Xie A, Wang H, Huang J, Sun M, Chen L. miR-3191 promotes the proliferation and metastasis of hepatocellular carcinoma via regulating PAK6. Infect Agent Cancer 2024; 19:64. [PMID: 39696440 DOI: 10.1186/s13027-024-00628-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 12/06/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND/AIMS microRNAs (miRNAs) contribute to tumorigenesis, progression and drug resistance of hepatocellular carcinoma (HCC). miR-3191 is a newly discovered miRNA, and its function and mechanism of action in biological processes and diseases are not completely understood. METHODS miR-3191 expression is determined via quantitative real-time polymerase chain reaction. Knockdown and overexpression of miR-3191 influence the proliferation and metastasis of HCC cells, which is measured by Cell Counting Kit-8 assay, Colony Formation assay and Cell metastasis assay. Protein expression is estimated by Western blot. The interplay between miR-3191 and target is validated by dual-luciferase reporter assay. RESULTS Here, we show that miR-3191 is upregulated in HCC tissues and associated with poor prognosis of HCC patients. Mechanistically, p21-activated protein kinase 6 (PAK6) was identified as a direct target of miR‑3191 in HCC. PAK6 knockdown partially recovered interference of miR‑3191‑induced decrease in cell proliferation and invasion. The accuracy of HCC patient prognosis could be improved by employing a combination of miR-3191 and PAK6 values. CONCLUSIONS miR-3191 promotes the proliferation and metastasis of HCC cells via targeting PAK6 and may serve as a prognostic biomarker and potential therapeutic target.
Collapse
Affiliation(s)
- Anqi Xie
- Community Health Service Center, Zhongshan Street, Songjiang District, Shanghai, China
| | - Hengjie Wang
- Department of Hepatic Surgery, Kunshan Hospital of Traditional Chinese Medicine, Kunshan, Jiangsu Province, 215300, China
| | - Jingchen Huang
- The National University of Malaysia, Kuala Lumpur, Malaysia
| | - Minmin Sun
- Department of Hepatic Surgery, Third Affiliated Hospital of Second Military Medical University, 225 Changhai Road, Shanghai, 200438, China.
| | - Lin Chen
- Community Health Service Center, Zhongshan Street, Songjiang District, Shanghai, China.
| |
Collapse
|
2
|
Pipili A, Babteen NA, Kuwair L, Jannet MB, Quist J, Ong KKV, Pitaluga R, Grigoriadis AG, Tutt A, Wells CM. PAK6 acts downstream of IQGAP3 to promote contractility in triple negative breast cancer cells. Cell Signal 2024; 121:111233. [PMID: 38763182 DOI: 10.1016/j.cellsig.2024.111233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 05/14/2024] [Accepted: 05/16/2024] [Indexed: 05/21/2024]
Abstract
Breast cancer is a heterogeneous disease that remains the most common malignancy among women worldwide. During genomic analysis of breast tumours, mRNA levels of IQGAP3 were found to be upregulated in triple negative tumours. IQGAP3 was subsequently found to be expressed across a panel of triple negative breast cancer (TNBC) cell lines. Depleting expression levels of IQGAP3 delivered elongated cells, disrupted cell migration, and inhibited the ability of cells to form specialised invasive adhesion structures, termed invadopodia. The morphological changes induced by IQGAP3 depletion were found to be dependent on RhoA. Indeed, reduced expression of IQGAP3 disrupted RhoA activity and actomyosin contractility. Interestingly, IQGAP3 was also found to interact with p-21 activated kinase 6 (PAK6); a protein already associated with the regulation of cell morphology. Moreover, PAK6 depletion phenocopied IQGAP3 depletion in these cells. Whereas PAK6 overexpression rescued the IQGAP3 depletion phenotype. Our work points to an important PAK6-IQGAP3-RhoA pathway that drives the cellular contractility of breast cancer cells promoting both cell migration and adhesive invasion of these cells. As this phenotype is relevant to the process of metastasis and re-seeding of metastasis, the pharmacological targeting of PAK6 could lead to clinical benefit in TNBC patients.
Collapse
Affiliation(s)
- Aikaterini Pipili
- School of Cancer and Pharmaceutical Sciences, Kings College London, London, UK
| | - Nouf A Babteen
- School of Cancer and Pharmaceutical Sciences, Kings College London, London, UK; Department of Biological Science, Collage of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Lujain Kuwair
- School of Cancer and Pharmaceutical Sciences, Kings College London, London, UK
| | - Mahfuja Bulu Jannet
- School of Cancer and Pharmaceutical Sciences, Kings College London, London, UK
| | - Jelmar Quist
- School of Cancer and Pharmaceutical Sciences, Kings College London, London, UK
| | - Karine K V Ong
- School of Cancer and Pharmaceutical Sciences, Kings College London, London, UK
| | - Ryan Pitaluga
- School of Cancer and Pharmaceutical Sciences, Kings College London, London, UK
| | - Anita G Grigoriadis
- School of Cancer and Pharmaceutical Sciences, Kings College London, London, UK
| | - Andrew Tutt
- School of Cancer and Pharmaceutical Sciences, Kings College London, London, UK; Tony Robins Breast Cancer Now Research Centre, The Institute of Cancer Research, London, UK
| | - Claire M Wells
- School of Cancer and Pharmaceutical Sciences, Kings College London, London, UK.
| |
Collapse
|
3
|
Huang Y, Bergant V, Grass V, Emslander Q, Hamad MS, Hubel P, Mergner J, Piras A, Krey K, Henrici A, Öllinger R, Tesfamariam YM, Dalla Rosa I, Bunse T, Sutter G, Ebert G, Schmidt FI, Way M, Rad R, Bowie AG, Protzer U, Pichlmair A. Multi-omics characterization of the monkeypox virus infection. Nat Commun 2024; 15:6778. [PMID: 39117661 PMCID: PMC11310467 DOI: 10.1038/s41467-024-51074-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 07/26/2024] [Indexed: 08/10/2024] Open
Abstract
Multiple omics analyzes of Vaccinia virus (VACV) infection have defined molecular characteristics of poxvirus biology. However, little is known about the monkeypox (mpox) virus (MPXV) in humans, which has a different disease manifestation despite its high sequence similarity to VACV. Here, we perform an in-depth multi-omics analysis of the transcriptome, proteome, and phosphoproteome signatures of MPXV-infected primary human fibroblasts to gain insights into the virus-host interplay. In addition to expected perturbations of immune-related pathways, we uncover regulation of the HIPPO and TGF-β pathways. We identify dynamic phosphorylation of both host and viral proteins, which suggests that MAPKs are key regulators of differential phosphorylation in MPXV-infected cells. Among the viral proteins, we find dynamic phosphorylation of H5 that influenced the binding of H5 to dsDNA. Our extensive dataset highlights signaling events and hotspots perturbed by MPXV, extending the current knowledge on poxviruses. We use integrated pathway analysis and drug-target prediction approaches to identify potential drug targets that affect virus growth. Functionally, we exemplify the utility of this approach by identifying inhibitors of MTOR, CHUK/IKBKB, and splicing factor kinases with potent antiviral efficacy against MPXV and VACV.
Collapse
Affiliation(s)
- Yiqi Huang
- Institute of Virology, Technical University of Munich, School of Medicine, Munich, Germany
| | - Valter Bergant
- Institute of Virology, Technical University of Munich, School of Medicine, Munich, Germany
| | - Vincent Grass
- Institute of Virology, Technical University of Munich, School of Medicine, Munich, Germany
| | - Quirin Emslander
- Institute of Virology, Technical University of Munich, School of Medicine, Munich, Germany
| | - M Sabri Hamad
- Institute of Virology, Technical University of Munich, School of Medicine, Munich, Germany
| | - Philipp Hubel
- Innate Immunity Laboratory, Max-Planck Institute of Biochemistry, Munich, Germany
- Core Facility Hohenheim, Universität Hohenheim, Stuttgart, Germany
| | - Julia Mergner
- Bavarian Center for Biomolecular Mass Spectrometry at University Hospital rechts der Isar (BayBioMS@MRI), Technical University of Munich, Munich, Germany
| | - Antonio Piras
- Institute of Virology, Technical University of Munich, School of Medicine, Munich, Germany
| | - Karsten Krey
- Institute of Virology, Technical University of Munich, School of Medicine, Munich, Germany
| | - Alexander Henrici
- Institute of Virology, Technical University of Munich, School of Medicine, Munich, Germany
| | - Rupert Öllinger
- Institute of Molecular Oncology and Functional Genomics and Department of Medicine II, School of Medicine, Technical University of Munich, Munich, Germany
| | - Yonas M Tesfamariam
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Ilaria Dalla Rosa
- Cellular signalling and cytoskeletal function laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Till Bunse
- Institute of Virology, Technical University of Munich, School of Medicine, Munich, Germany
| | - Gerd Sutter
- Institute for Infectious Diseases and Zoonoses, Department of Veterinary Sciences, LMU Munich, Munich, Germany
- German Centre for Infection Research (DZIF), Partner site Munich, Munich, Germany
| | - Gregor Ebert
- Institute of Virology, Technical University of Munich, School of Medicine/Helmholtz Munich, Munich, Germany
| | - Florian I Schmidt
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Michael Way
- Cellular signalling and cytoskeletal function laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
- Department of Infectious Disease, Imperial College, London, UK
| | - Roland Rad
- Institute of Molecular Oncology and Functional Genomics and Department of Medicine II, School of Medicine, Technical University of Munich, Munich, Germany
| | - Andrew G Bowie
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Ulrike Protzer
- German Centre for Infection Research (DZIF), Partner site Munich, Munich, Germany
- Institute of Virology, Technical University of Munich, School of Medicine/Helmholtz Munich, Munich, Germany
| | - Andreas Pichlmair
- Institute of Virology, Technical University of Munich, School of Medicine, Munich, Germany.
- German Centre for Infection Research (DZIF), Partner site Munich, Munich, Germany.
| |
Collapse
|
4
|
Huang H, Wang S, Guan Y, Ren J, Liu X. Molecular basis and current insights of atypical Rho small GTPase in cancer. Mol Biol Rep 2024; 51:141. [PMID: 38236467 DOI: 10.1007/s11033-023-09140-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 12/11/2023] [Indexed: 01/19/2024]
Abstract
Atypical Rho GTPases are a subtype of the Rho GTPase family that are involved in diverse cellular processes. The typical Rho GTPases, led by RhoA, Rac1 and Cdc42, have been well studied, while relative studies on atypical Rho GTPases are relatively still limited and have great exploration potential. With the increase in studies, current evidence suggests that atypical Rho GTPases regulate multiple biological processes and play important roles in the occurrence and development of human cancers. Therefore, this review mainly discusses the molecular basis of atypical Rho GTPases and their roles in cancer. We summarize the sequence characteristics, subcellular localization and biological functions of each atypical Rho GTPase. Moreover, we review the recent advances and potential mechanisms of atypical Rho GTPases in the development of multiple cancers. A comprehensive understanding and extensive exploration of the biological functions of atypical Rho GTPases and their molecular mechanisms in tumors will provide important insights into the pathophysiology of tumors and the development of cancer therapeutic strategies.
Collapse
Affiliation(s)
- Hua Huang
- Center of Excellence for Environmental Safety and Biological Effects, Faculty of Environment and Life, Beijing International Science and Technology Cooperation Base for Antiviral Drugs, Beijing University of Technology, Beijing, 100124, China
| | - Sijia Wang
- Center of Excellence for Environmental Safety and Biological Effects, Faculty of Environment and Life, Beijing International Science and Technology Cooperation Base for Antiviral Drugs, Beijing University of Technology, Beijing, 100124, China
| | - Yifei Guan
- Center of Excellence for Environmental Safety and Biological Effects, Faculty of Environment and Life, Beijing International Science and Technology Cooperation Base for Antiviral Drugs, Beijing University of Technology, Beijing, 100124, China
| | - Jing Ren
- Department of Plastic and Reconstructive Surgery, The First Medical Center, Chinese PLA (People's Liberation Army) General Hospital, Beijing, 100853, China.
| | - Xinhui Liu
- Center of Excellence for Environmental Safety and Biological Effects, Faculty of Environment and Life, Beijing International Science and Technology Cooperation Base for Antiviral Drugs, Beijing University of Technology, Beijing, 100124, China.
- Faculty of Environment and Life, Beijing University of Technology, Beijing, 100124, China.
| |
Collapse
|
5
|
Juan-Guadarrama DG, Beltrán-Navarro YM, Reyes-Cruz G, Vázquez-Prado J. Ephexin3/ARHGEF5 Together with Cell Migration Signaling Partners within the Tumor Microenvironment Define Prognostic Transcriptional Signatures in Multiple Cancer Types. Int J Mol Sci 2023; 24:16427. [PMID: 38003617 PMCID: PMC10671824 DOI: 10.3390/ijms242216427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/08/2023] [Accepted: 11/15/2023] [Indexed: 11/26/2023] Open
Abstract
Cancer cell migration involves a repertoire of signaling proteins that lead cytoskeleton reorganization as a critical step in metastatic dissemination. RhoGEFs are multidomain effectors that integrate signaling inputs to activate the molecular switches that orchestrate actin cytoskeleton reorganization. Ephexins, a group of five RhoGEFs, play oncogenic roles in invasive and metastatic cancer, leading to a mechanistic hypothesis about their function as signaling nodes assembling functional complexes that guide cancer cell migration. To identify clinically significant Ephexin signaling partners, we applied three systematic data mining strategies, based on the screening of essential Ephexins in multiple cancer cell lines and the identification of coexpressed signaling partners in the TCGA cancer patient datasets. Based on the domain architecture of encoded proteins and gene ontology criteria, we selected Ephexin signaling partners with a role in cytoskeletal reorganization and cell migration. We focused on Ephexin3/ARHGEF5, identified as an essential gene in multiple cancer cell types. Based on significant coexpression data and coessentiality, the signaling repertoire that accompanies Ephexin3 corresponded to three groups: pan-cancer, cancer-specific and coessential. To further select the Ephexin3 signaling partners likely to be relevant in clinical settings, we first identified those whose high expression was statistical linked to shorter patient survival. The resulting Ephexin3 transcriptional signatures represent significant accumulated risk, predictive of shorter survival, in 17 cancer types, including PAAD, LUAD, LGG, OSC, AML, KIRC, THYM, BLCA, LIHC and UCEC. The signaling landscape that accompanies Ephexin3 in various cancer types included the tyrosine kinase receptor MET and the tyrosine phosphatase receptor PTPRF, the serine/threonine kinases MARK2 and PAK6, the Rho GTPases RHOD, RHOF and RAC1, and the cytoskeletal regulator DIAHP1. Our findings set the basis to further explore the role of Ephexin3/ARHGEF5 as an essential effector and signaling hub in cancer cell migration.
Collapse
Affiliation(s)
- Dante Gustavo Juan-Guadarrama
- Department of Pharmacology, Cinvestav-IPN, Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, Mexico City 07360, Mexico
| | - Yarely Mabell Beltrán-Navarro
- Department of Pharmacology, Cinvestav-IPN, Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, Mexico City 07360, Mexico
| | - Guadalupe Reyes-Cruz
- Department of Cell Biology, Cinvestav-IPN, Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, Mexico City 07360, Mexico
| | - José Vázquez-Prado
- Department of Pharmacology, Cinvestav-IPN, Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, Mexico City 07360, Mexico
| |
Collapse
|
6
|
Cai X, Yang R, Shi W, Cai Y, Ma Z. Exploration of the common pathogenic link between COVID-19 and diabetic foot ulcers: An in silico approach. Health Sci Rep 2023; 6:e1686. [PMID: 37936615 PMCID: PMC10626003 DOI: 10.1002/hsr2.1686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 10/09/2023] [Accepted: 10/23/2023] [Indexed: 11/09/2023] Open
Abstract
Background and Aims The Coronavirus Disease-19 (COVID-19) is posing an ongoing threat to human health. Patients of diabetic foot ulcer (DFU) are susceptible to COVID-19-induced adverse outcomes. Nevertheless, investigations into their mutual molecular mechanisms have been limited to date. In the present work, we tried to uncover the shared pathogenesis and regulatory gene targets of COVID-19 and DFU. Methods In this study, we chose GSE161281 as the COVID-19 data set, which contained severe acute respiratory syndrome coronavirus 2 infected human induced embryonic stem cell-derived peripheral neurons (n = 2) with uninfected controls (n = 2). The GSE134431 designated as the DFU data set, comprising full-thickness DFU (n = 13) and diabetic foot skin (n = 8) samples from diabetic patients. The differential expressed genes (DEGs) were identified from GSE161281 and GSE134431, and the common DEGs between COVID-19 and DFU were extracted. Multifactor regulatory network and co-expression network of the common DEGs were analyzed, along with candidate drug prediction. Results Altogether, six common DEGs (dickkopf-related protein 1 [DKK1], serine proteinase inhibitor A3 [SERPINA3], ras homolog family member D [RHOD], myelin protein zero like 3 [MPZL3], Claudin-11 [CLDN11], and epidermal growth factor receptor pathway substrate 8-like 1 [EPS8L1]) were found between COVID-19 and DFU. Functional analyses indicated that pathways of apoptotic and Wnt signaling may contribute to progression of COVID-19. Gene co-expression network implied the shared pathways of immune regulation and cytokine response participated collectively in the development of DFU and COVID-19. A multifactor regulatory network was constructed integrating the corresponding microRNAs (miRNAs) and transcription factors. Additionally, we proposed potential drug objects for the combined therapy. Conclusion Our study revealed the shared molecular mechanisms underlying COVID-19 and DFU. The identified pivotal targets and common pathways can provide new perspectives for further research and assist the development of management strategies in patients of DFU complicated with COVID-19.
Collapse
Affiliation(s)
- Xueyao Cai
- Department of Burn and Plastic SurgeryDongguan Tungwah HospitalDongguanChina
| | - Ruijin Yang
- Department of Burn and Plastic SurgeryDongguan Tungwah HospitalDongguanChina
| | - Wenjun Shi
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yuchen Cai
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Zhengzheng Ma
- Department of Burn and Plastic SurgeryDongguan Tungwah HospitalDongguanChina
| |
Collapse
|
7
|
Gao YN, Min L, Yang X, Wang JQ, Zheng N. The coexistence of aflatoxin M1 and ochratoxin A induced intestinal barrier disruption via the regulation of key differentially expressed microRNAs and long non-coding RNAs in BALB/c mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 264:115428. [PMID: 37688864 DOI: 10.1016/j.ecoenv.2023.115428] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 08/07/2023] [Accepted: 08/30/2023] [Indexed: 09/11/2023]
Abstract
Food safety can be seriously threatened by the existence of both aflatoxin M1 (AFM1) and ochratoxin A (OTA) in milk and corresponding products. The importance of intestine integrity in preserving human health is widely understood in vitro, but the fundamental processes by which AFM1 and OTA cause disruption of the intestinal barrier are as yet unknown, especially in vivo. Based on the analysis of the whole transcriptome of BALB/c mice, the competing endogenous RNA (ceRNA) regulation network was obtained in the current study. Each of 12 mice were separated into five treatments: saline solution treatment, 1.0% DMSO vehicle control treatment, 3.0 mg/kg b.w. individual AFM1 treatment (AFM1), 3.0 mg/kg b.w. individual OTA treatment (OTA), and combined mycotoxins treatment (AFM1 +OTA). The study period lasted 28 days. The jejunum tissue was collected for the histological assessment and whole transcriptome analysis, and the whole blood was collected, and determination of serum biochemical indicators. The phenotypic results demonstrated that AFM1 and OTA caused intestinal barrier disruption via an increased apoptosis level and decreased expression of tight junction (TJ) proteins. The ceRNA network demonstrated that AFM1 and OTA induced cell apoptosis through activating the expression of DUSP9 and suppressing the expression of PLA2G2D, which were regulated by differentially expressed microRNAs (DEmiRNAs) (miR-124-y, miR-194-z, miR-224-x, and miR-452-x) and differentially expressed long non-coding RNAs (DElncRNAs) (FUT8 and GPR31C). And AFM1 and OTA decreased TJ proteins via inhibiting the expression of PAK6, which was regulated by several important DEmiRNAs and DElncRNAs. These DE RNAs in intestinal integrity were involved in MAPK and Ras signaling pathway. Overall, our findings expand the current knowledge regarding the potential mechanisms of intestinal integrity disruption brought on by AFM1 and OTA in vivo.
Collapse
Affiliation(s)
- Ya-Nan Gao
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Li Min
- Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Public Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
| | - Xue Yang
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Jia-Qi Wang
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Nan Zheng
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| |
Collapse
|
8
|
Lu J, Liu W, Chen XZ, Wang Y, Ying T, Qiao L, Liu YJ, Liu B. Temporal proteomic profiling reveals functional pathways in vaccinia virus-induced cell migration. Front Microbiol 2023; 14:1185960. [PMID: 37303799 PMCID: PMC10249495 DOI: 10.3389/fmicb.2023.1185960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 04/03/2023] [Indexed: 06/13/2023] Open
Abstract
Introduction Viral diseases have always been intricate and persistent issues throughout the world and there is a lack of holistic discoveries regarding the molecular dysregulations of virus-host interactions. The temporal proteomics strategy can identify various differentially expressed proteins and offer collaborated interaction networks under pathological conditions. Method Herein, temporal proteomics at various hours post infection of Vero cells were launched to uncover molecular alternations during vaccinia virus (VACV)-induced cell migration. Different stages of infection were included to differentiate gene ontologies and critical pathways at specific time points of infection via bioinformatics. Results Bioinformatic results showed functional and distinct ontologies and pathways at different stages of virus infection. The enrichment of interaction networks and pathways verified the significances of the regulation of actin cytoskeleton and lamellipodia during VACV-induced fast cell motility. Discussion The current results offer a systematic proteomic profiling of molecular dysregulations at different stages of VACV infection and potential biomedical targets for treating viral diseases.
Collapse
Affiliation(s)
- Jiayin Lu
- Department of Chemistry, Shanghai Stomatological Hospital, Institutes of Biomedical Sciences, Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), State Key Lab of Molecular Engineering of Polymers, Fudan University, Shanghai, China
| | - Wei Liu
- Department of Chemistry, Shanghai Stomatological Hospital, Institutes of Biomedical Sciences, Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), State Key Lab of Molecular Engineering of Polymers, Fudan University, Shanghai, China
| | - Xue-Zhu Chen
- Department of Chemistry, Shanghai Stomatological Hospital, Institutes of Biomedical Sciences, Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), State Key Lab of Molecular Engineering of Polymers, Fudan University, Shanghai, China
| | - Yiwen Wang
- Department of Chemistry, Shanghai Stomatological Hospital, Institutes of Biomedical Sciences, Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), State Key Lab of Molecular Engineering of Polymers, Fudan University, Shanghai, China
| | - Tianlei Ying
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Liang Qiao
- Department of Chemistry, Shanghai Stomatological Hospital, Institutes of Biomedical Sciences, Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), State Key Lab of Molecular Engineering of Polymers, Fudan University, Shanghai, China
| | - Yan-Jun Liu
- Department of Chemistry, Shanghai Stomatological Hospital, Institutes of Biomedical Sciences, Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), State Key Lab of Molecular Engineering of Polymers, Fudan University, Shanghai, China
| | - Baohong Liu
- Department of Chemistry, Shanghai Stomatological Hospital, Institutes of Biomedical Sciences, Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), State Key Lab of Molecular Engineering of Polymers, Fudan University, Shanghai, China
| |
Collapse
|
9
|
Hernandez-Gonzalez M, Calcraft T, Nans A, Rosenthal PB, Way M. A succession of two viral lattices drives vaccinia virus assembly. PLoS Biol 2023; 21:e3002005. [PMID: 36862727 PMCID: PMC10013923 DOI: 10.1371/journal.pbio.3002005] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 03/14/2023] [Accepted: 01/19/2023] [Indexed: 03/03/2023] Open
Abstract
During its cytoplasmic replication, vaccinia virus assembles non-infectious spherical immature virions (IV) coated by a viral D13 lattice. Subsequently, IV mature into infectious brick-shaped intracellular mature virions (IMV) that lack D13. Here, we performed cryo-electron tomography (cryo-ET) of frozen-hydrated vaccinia-infected cells to structurally characterise the maturation process in situ. During IMV formation, a new viral core forms inside IV with a wall consisting of trimeric pillars arranged in a new pseudohexagonal lattice. This lattice appears as a palisade in cross-section. As maturation occurs, which involves a 50% reduction in particle volume, the viral membrane becomes corrugated as it adapts to the newly formed viral core in a process that does not appear to require membrane removal. Our study suggests that the length of this core is determined by the D13 lattice and that the consecutive D13 and palisade lattices control virion shape and dimensions during vaccinia assembly and maturation.
Collapse
Affiliation(s)
- Miguel Hernandez-Gonzalez
- Cellular signalling and cytoskeletal function laboratory, The Francis Crick Institute, London, United Kingdom
| | - Thomas Calcraft
- Structural Biology of Cells and Viruses Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Andrea Nans
- Structural Biology Science Technology Platform, The Francis Crick Institute, London, United Kingdom
| | - Peter B Rosenthal
- Structural Biology of Cells and Viruses Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Michael Way
- Cellular signalling and cytoskeletal function laboratory, The Francis Crick Institute, London, United Kingdom
- Department of Infectious Disease, Imperial College, London, United Kingdom
- * E-mail:
| |
Collapse
|
10
|
Chetty AK, Ha BH, Boggon TJ. Rho family GTPase signaling through type II p21-activated kinases. Cell Mol Life Sci 2022; 79:598. [PMID: 36401658 PMCID: PMC10105373 DOI: 10.1007/s00018-022-04618-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 10/07/2022] [Accepted: 10/28/2022] [Indexed: 11/21/2022]
Abstract
Signaling from the Rho family small GTPases controls a wide range of signaling outcomes. Key among the downstream effectors for many of the Rho GTPases are the p21-activated kinases, or PAK group. The PAK family comprises two types, the type I PAKs (PAK1, 2 and 3) and the type II PAKs (PAK4, 5 and 6), which have distinct structures and mechanisms of regulation. In this review, we discuss signal transduction from Rho GTPases with a focus on the type II PAKs. We discuss the role of PAKs in signal transduction pathways and selectivity of Rho GTPases for PAK family members. We consider the less well studied of the Rho GTPases and their PAK-related signaling. We then discuss the molecular basis for kinase domain recognition of substrates and for regulation of signaling. We conclude with a discussion of the role of PAKs in cross talk between Rho family small GTPases and the roles of PAKs in disease.
Collapse
Affiliation(s)
- Ashwin K Chetty
- Yale College, New Haven, CT, 06520, USA
- Department of Molecular Biophysics and Biochemistry, Yale University, 333 Cedar Street, New Haven, CT, 06520, USA
| | - Byung Hak Ha
- Department of Pharmacology, Yale University, 333 Cedar Street, New Haven, CT, 06520, USA
| | - Titus J Boggon
- Department of Molecular Biophysics and Biochemistry, Yale University, 333 Cedar Street, New Haven, CT, 06520, USA.
- Department of Pharmacology, Yale University, 333 Cedar Street, New Haven, CT, 06520, USA.
- Yale Cancer Center, Yale University, 333 Cedar Street, New Haven, CT, 06520, USA.
| |
Collapse
|
11
|
Matozo T, Kogachi L, de Alencar BC. Myosin motors on the pathway of viral infections. Cytoskeleton (Hoboken) 2022; 79:41-63. [PMID: 35842902 DOI: 10.1002/cm.21718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 06/25/2022] [Accepted: 07/07/2022] [Indexed: 01/30/2023]
Abstract
Molecular motors are microscopic machines that use energy from adenosine triphosphate (ATP) hydrolysis to generate movement. While kinesins and dynein are molecular motors associated with microtubule tracks, myosins bind to and move on actin filaments. Mammalian cells express several myosin motors. They power cellular processes such as endo- and exocytosis, intracellular trafficking, transcription, migration, and cytokinesis. As viruses navigate through cells, they may take advantage or be hindered by host components and machinery, including the cytoskeleton. This review delves into myosins' cell roles and compares them to their reported functions in viral infections. In most cases, the previously described myosin functions align with their reported role in viral infections, although not in all cases. This opens the possibility that knowledge obtained from studying myosins in viral infections might shed light on new physiological roles for myosins in cells. However, given the high number of myosins expressed and the variety of viruses investigated in the different studies, it is challenging to infer whether the interactions found are specific to a single virus or can be applied to other viruses with the same characteristics. We conclude that the participation of myosins in viral cycles is still a largely unexplored area, especially concerning unconventional myosins.
Collapse
Affiliation(s)
- Tais Matozo
- Departamento de Imunologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Leticia Kogachi
- Departamento de Imunologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Bruna Cunha de Alencar
- Departamento de Imunologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
12
|
β-Arrestin2 Is Critically Involved in the Differential Regulation of Phosphosignaling Pathways by Thyrotropin-Releasing Hormone and Taltirelin. Cells 2022; 11:cells11091473. [PMID: 35563779 PMCID: PMC9103620 DOI: 10.3390/cells11091473] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 04/22/2022] [Accepted: 04/25/2022] [Indexed: 12/17/2022] Open
Abstract
In recent years, thyrotropin-releasing hormone (TRH) and its analogs, including taltirelin (TAL), have demonstrated a range of effects on the central nervous system that represent potential therapeutic agents for the treatment of various neurological disorders, including neurodegenerative diseases. However, the molecular mechanisms of their actions remain poorly understood. In this study, we investigated phosphosignaling dynamics in pituitary GH1 cells affected by TRH and TAL and the putative role of β-arrestin2 in mediating these effects. Our results revealed widespread alterations in many phosphosignaling pathways involving signal transduction via small GTPases, MAP kinases, Ser/Thr- and Tyr-protein kinases, Wnt/β-catenin, and members of the Hippo pathway. The differential TRH- or TAL-induced phosphorylation of numerous proteins suggests that these ligands exhibit some degree of biased agonism at the TRH receptor. The different phosphorylation patterns induced by TRH or TAL in β-arrestin2-deficient cells suggest that the β-arrestin2 scaffold is a key factor determining phosphorylation events after TRH receptor activation. Our results suggest that compounds that modulate kinase and phosphatase activity can be considered as additional adjuvants to enhance the potential therapeutic value of TRH or TAL.
Collapse
|
13
|
Lou Y, Jiang Y, Liang Z, Liu B, Li T, Zhang D. Role of RhoC in cancer cell migration. Cancer Cell Int 2021; 21:527. [PMID: 34627249 PMCID: PMC8502390 DOI: 10.1186/s12935-021-02234-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 09/27/2021] [Indexed: 12/20/2022] Open
Abstract
Migration is one of the five major behaviors of cells. Although RhoC-a classic member of the Rho gene family-was first identified in 1985, functional RhoC data have only been widely reported in recent years. Cell migration involves highly complex signaling mechanisms, in which RhoC plays an essential role. Cell migration regulated by RhoC-of which the most well-known function is its role in cancer metastasis-has been widely reported in breast, gastric, colon, bladder, prostate, lung, pancreatic, liver, and other cancers. Our review describes the role of RhoC in various types of cell migration. The classic two-dimensional cell migration cycle constitutes cell polarization, adhesion regulation, cell contraction and tail retraction, most of which are modulated by RhoC. In the three-dimensional cell migration model, amoeboid migration is the most classic and well-studied model. Here, RhoC modulates the formation of membrane vesicles by regulating myosin II, thereby affecting the rate and persistence of amoeba-like migration. To the best of our knowledge, this review is the first to describe the role of RhoC in all cell migration processes. We believe that understanding the detail of RhoC-regulated migration processes will help us better comprehend the mechanism of cancer metastasis. This will contribute to the study of anti-metastatic treatment approaches, aiding in the identification of new intervention targets for therapeutic or genetic transformational purposes.
Collapse
Affiliation(s)
- Yingyue Lou
- Department of Plastic and Reconstructive Surgery, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yuhan Jiang
- Department of Plastic and Reconstructive Surgery, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Zhen Liang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Bingzhang Liu
- Department of Plastic and Reconstructive Surgery, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Tian Li
- Department of Plastic and Reconstructive Surgery, The First Hospital of Jilin University, Changchun, Jilin, China.
| | - Duo Zhang
- Department of Plastic and Reconstructive Surgery, The First Hospital of Jilin University, Changchun, Jilin, China.
| |
Collapse
|
14
|
The actin nucleation factors JMY and WHAMM enable a rapid Arp2/3 complex-mediated intrinsic pathway of apoptosis. PLoS Genet 2021; 17:e1009512. [PMID: 33872315 PMCID: PMC8084344 DOI: 10.1371/journal.pgen.1009512] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 04/29/2021] [Accepted: 03/28/2021] [Indexed: 01/02/2023] Open
Abstract
The actin cytoskeleton is a well-known player in most vital cellular processes, but comparably little is understood about how the actin assembly machinery impacts programmed cell death pathways. In the current study, we explored roles for the human Wiskott-Aldrich Syndrome Protein (WASP) family of actin nucleation factors in DNA damage-induced apoptosis. Inactivation of each WASP-family gene revealed that two of them, JMY and WHAMM, are necessary for rapid apoptotic responses. JMY and WHAMM participate in a p53-dependent cell death pathway by enhancing mitochondrial permeabilization, initiator caspase cleavage, and executioner caspase activation. JMY-mediated apoptosis requires actin nucleation via the Arp2/3 complex, and actin filaments are assembled in cytoplasmic territories containing clusters of cytochrome c and active caspase-3. The loss of JMY additionally results in significant changes in gene expression, including upregulation of the WHAMM-interacting G-protein RhoD. Depletion or deletion of RHOD increases cell death, suggesting that RhoD normally contributes to cell survival. These results give rise to a model in which JMY and WHAMM promote intrinsic cell death responses that can be opposed by RhoD.
Collapse
|
15
|
Zheng J, Zhang C, Li Y, Jiang Y, Xing B, Du X. p21-activated kinase 6 controls mitosis and hepatocellular carcinoma progression by regulating Eg5. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1868:118888. [PMID: 33098954 DOI: 10.1016/j.bbamcr.2020.118888] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 10/07/2020] [Accepted: 10/09/2020] [Indexed: 02/07/2023]
Abstract
P21-activated kinases 6 (PAK6) associated with many fundamental cellular processes in cancer including cell-cell adhesion, migration and apoptosis. Here, we report a novel function of PAK6 in mitosis. Expression of PAK6 peaks in the M phase. Knockdown of PAK6 increases cell number in G2/M and promotes cell proliferation. PAK6 specifically colocalizes with Eg5 in the centrosome. Depletion of PAK6 results in multipolar spindle and a simultaneous upregulation of Eg5. Further, the PAK6 depletion-induced multiple spindle and cell cycle progression is reversed by knockdown of Eg5. These data suggest that PAK6 regulates spindle formation and cell cycle by regulating Eg5 expression. Additionally, expression of PAK6 is upregulated when Eg5 is downregulated or inhibited. Thus, PAK6 and Eg5 negatively inter-regulate each other. Significantly, the effect of PAK6 expression on the outcome of the HCC patients is controlled by Eg5 expression. Inhibition of Eg5 reverses PAK6 depletion-promoted cell invasion. Collectively, our data indicate that the inter-regulation between PAK6 and Eg5 might promote the progression of HCC.
Collapse
Affiliation(s)
- Jiaojiao Zheng
- Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100083, China
| | - Chunfeng Zhang
- Department of Medical Genetics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100083, China
| | - Yuan Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Hepatopancreatobiliary Surgery Department I, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Yang Jiang
- Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100083, China
| | - Baocai Xing
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Hepatopancreatobiliary Surgery Department I, Peking University Cancer Hospital & Institute, Beijing 100142, China.
| | - Xiaojuan Du
- Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100083, China.
| |
Collapse
|
16
|
Kelkar M, Bohec P, Charras G. Mechanics of the cellular actin cortex: From signalling to shape change. Curr Opin Cell Biol 2020; 66:69-78. [DOI: 10.1016/j.ceb.2020.05.008] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 03/30/2020] [Accepted: 05/08/2020] [Indexed: 01/17/2023]
|
17
|
Sundararaman A, Mellor H. A functional antagonism between RhoJ and Cdc42 regulates fibronectin remodelling during angiogenesis. Small GTPases 2020; 12:241-245. [PMID: 32857689 PMCID: PMC8205010 DOI: 10.1080/21541248.2020.1809927] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Angiogenesis is the formation of new blood vessels from pre-existing ones. Angiogenesis requires endothelial cells to change shape and polarity, as well as acquire the ability to directionally migrate ‒ processes that are classically regulated by the Rho family of GTPases. RhoJ (previously TCL) is an endothelium enriched Rho GTPase with a 78% amino acid similarity to the ubiquitously expressed Cdc42. In our recent publication, we demonstrate that α5β1 integrin co-traffics with RhoJ. RhoJ specifically represses the internalization of the active α5β1 conformer, leading to a reduced ability of endothelial cells to form fibronectin fibrils. Surprisingly, this function of RhoJ is in opposition to the role of Cdc42, a known driver of fibrillogenesis. Intriguingly, we discovered that the competition for limiting amounts of the shared effector, PAK3, could explain the ability of these two Rho GTPases to regulate fibrillogenesis in opposing directions. Consequently, RhoJ null mice show excessive fibronectin deposition around retinal vessels, possibly due to the unopposed action of Cdc42. Our work suggests that the functional antagonism between RhoJ and Cdc42 could restrict fibronectin remodelling to sites of active angiogenesis to form a provisional matrix for vessel growth. One correlate of our findings is that RhoJ dependent repression of fibronectin remodelling could be atheroprotective in quiescent vessels.
Collapse
Affiliation(s)
- Ananthalakshmy Sundararaman
- Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, Kerala, India
| | - Harry Mellor
- School of Biochemistry, Biomedical Sciences Building, University of Bristol, Bristol, UK
| |
Collapse
|
18
|
Li T, Li Y, Liu T, Hu B, Li J, Liu C, Liu T, Li F. Mitochondrial PAK6 inhibits prostate cancer cell apoptosis via the PAK6-SIRT4-ANT2 complex. Theranostics 2020; 10:2571-2586. [PMID: 32194820 PMCID: PMC7052886 DOI: 10.7150/thno.42874] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 01/11/2020] [Indexed: 12/18/2022] Open
Abstract
Rationale: P21-activated kinase 6 (PAK6) is a member of the class II PAKs family, which is a conserved family of serine/threonine kinases. Although the effects of PAK6 on many malignancies, especially in prostate cancer, have been studied for a long time, the role of PAK6 in mitochondria remains unknown. Methods: The expression of PAK6, SIRT4 and ANT2 in prostate cancer and adjacent non-tumor tissues was detected by immunohistochemistry. Immunofuorescence and immunoelectron microscopy were used to determine the subcellular localization of PAK6. Immunoprecipitation, immunofuorescence and ubiquitination assays were performed to determine how PAK6 regulates SIRT4, how SIRT4 regulates ANT2, and how PAK6 regulates ANT2. Flow cytometry detection and xenograft models were used to evaluate the impact of ANT2 mutant expression on the prostate cancer cell cycle and apoptosis regulation. Results: The present study revealed that the PAK6-SIRT4-ANT2 complex is involved in mitochondrial apoptosis in prostate cancer cells. It was found that PAK6 is mainly located in the mitochondrial inner membrane, in which PAK6 promotes SIRT4 ubiquitin-mediated proteolysis. Furthermore, SIRT4 deprives the ANT2 acetylation at K105 to promote its ubiquitination degradation. Hence, PAK6 adjusts the acetylation level of ANT2 through the PAK6-SIRT4-ANT2 pathway, in order to regulate the stability of ANT2. Meanwhile, PAK6 directly phosphorylates ANT2 atT107 to inhibit the apoptosis of prostate cancer cells. Therefore, the phosphorylation and deacetylation modifications of ANT2 are mutually regulated, leading to tumor growth in vivo. Consistently, these clinical prostate cancer tissue evaluations reveal that PAK6 is positively correlated with ANT2 expression, but negatively correlated with SIRT4. Conclusion: These present findings suggest the pivotal role of the PAK6-SIRT4-ANT2 complex in the apoptosis of prostate cancer. This complex could be a potential biomarker for the treatment and prognosis of prostate cancer.
Collapse
Affiliation(s)
- Tingting Li
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the PRC, and Key Laboratory of Medical Cell Biology, Ministry of Education of the PRC, Shenyang 110122, Liaoning, China
| | - Yang Li
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the PRC, and Key Laboratory of Medical Cell Biology, Ministry of Education of the PRC, Shenyang 110122, Liaoning, China
| | - Tong Liu
- Medical Research Center, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China
| | - Bingtao Hu
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the PRC, and Key Laboratory of Medical Cell Biology, Ministry of Education of the PRC, Shenyang 110122, Liaoning, China
| | - Jiabin Li
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the PRC, and Key Laboratory of Medical Cell Biology, Ministry of Education of the PRC, Shenyang 110122, Liaoning, China
| | - Chen Liu
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the PRC, and Key Laboratory of Medical Cell Biology, Ministry of Education of the PRC, Shenyang 110122, Liaoning, China
| | - Tao Liu
- Department of Urology, the First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning, China
| | - Feng Li
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the PRC, and Key Laboratory of Medical Cell Biology, Ministry of Education of the PRC, Shenyang 110122, Liaoning, China
| |
Collapse
|
19
|
Thomas P, Pranatharthi A, Ross C, Srivastava S. RhoC: a fascinating journey from a cytoskeletal organizer to a Cancer stem cell therapeutic target. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:328. [PMID: 31340863 PMCID: PMC6651989 DOI: 10.1186/s13046-019-1327-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 07/15/2019] [Indexed: 01/05/2023]
Abstract
Tumor heterogeneity results in differential response to therapy due to the existence of plastic tumor cells, called cancer stem cells (CSCs), which exhibit the property of resistance to therapy, invasion and metastasis. These cells have a distinct, signaling network active at every stage of progression. It is difficult to envisage that the CSCs will have a unique set of signaling pathways regulating every stage of disease progression. Rather, it would be easier to believe that a single pivotal pathway having significant contribution at every stage, which can further turn on a battery of signaling mechanisms specific to that stage, would be instrumental in regulating the signaling network, enabling easy transition from one state to another. In this context, we discuss the role of RhoC which has contributed to several phenotypes during tumor progression. RhoC (Ras homolog gene family member C) has been widely reported to regulate actin organization. It has been shown to impact the motility of cancer cells, resultantly affecting invasion and metastasis, and has contributed to carcinoma progression of the breast, pancreas, lung, ovaries and cervix, among several others. The most interesting finding has been its indispensable role in metastasis. Also, it has the ability to modulate various other phenotypes like angiogenesis, motility, invasion, metastasis, and anoikis resistance. These observations suggest that RhoC imparts the plasticity required by tumor cells to exhibit such diverse functions based on microenvironmental cues. This was further confirmed by recent reports which show that it regulates cancer stem cells in breast, ovary and head and neck cancers. Studies also suggest that the inhibition of RhoC results in abolition of advanced tumor phenotypes. Our review throws light on how RhoC, which is capable of modulating various phenotypes may be the apt core signaling candidate regulating disease progression. Additionally, mice studies show that RhoC is not essential for embryogenesis, giving scope for its development as a possible therapeutic target. This review thus stresses on the need to understand the protein and its functioning in greater detail to enable its development as a stem cell marker and a possible therapeutic target.
Collapse
Affiliation(s)
- Pavana Thomas
- Translational and Molecular Biology Laboratory (TMBL), St. John's Research Institute (SJRI), Bangalore, 560034, India.,School of Integrative Health Sciences, The University of Trans-Disciplinary Health Sciences and Technology (TDU), Bangalore, 560064, India
| | - Annapurna Pranatharthi
- Rajiv Gandhi University of Health Sciences (RGUHS), Bangalore, 560041, India.,National Centre for Biological Sciences (NCBS), Bangalore, 560065, India.,Translational and Molecular Biology Laboratory (TMBL), Department of Medicine, St. John's Medical College Hospital (SJMCH), Bangalore, 560034, India
| | - Cecil Ross
- Translational and Molecular Biology Laboratory (TMBL), Department of Medicine, St. John's Medical College Hospital (SJMCH), Bangalore, 560034, India
| | - Sweta Srivastava
- Translational and Molecular Biology Laboratory (TMBL), Department of Transfusion Medicine and Immunohematology, St. John's Medical College Hospital (SJMCH), Bangalore, 560034, India.
| |
Collapse
|
20
|
Lu WJ, Gao FX, Wang Y, Zhang QY, Li Z, Zhang XJ, Zhou L, Gui JF. Differential expression of innate and adaptive immune genes in the survivors of three gibel carp gynogenetic clones after herpesvirus challenge. BMC Genomics 2019; 20:432. [PMID: 31138127 PMCID: PMC6540555 DOI: 10.1186/s12864-019-5777-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 05/07/2019] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Accompanied with rapid growth and high density aquaculture, gibel carp has been seriously threatened by Carassius auratus herpesvirus (CaHV) since 2012. In previous study, distinct CaHV resistances and immune responses were revealed in the diseased individuals of three gibel carp gynogenetic clones (A+, F and H). However, little is known about the gene expression changes in the survivors after CaHV challenge, particularly their differences of innate and adaptive immune system between susceptible clone and resistant clone. RESULTS We firstly confirmed the CaHV carrier state in the survivors of three gibel carp clones after CaHV challenge by evaluating the abundances of five CaHV genes. The assay of viral loads indicated the resistant clone H possessed not only stronger resistance but also higher tolerance to CaHV. Then, 2818, 4047 and 3323 differentially expressed unigenes (DEUs) were screened from the head-kidney transcriptome profiles of survivors compared with controls from clone A+, F and H. GO and KEGG analysis suggested that a persistent immune response might sustain in resistant clone H and F, while susceptible clone A+ had a long-term impact on the circulatory system which was consistent with the major symptoms of bleeding caused by CaHV. Among the top 30 enriched pathways of specifically up-regulated DEUs in respective clones, 26, 7 and 15 pathways in clone H, F and A+ were associated with infections, diseases, or immune-related pathways respectively. In addition, 20 pathways in clone F belonged to "metabolism" or "biogenesis", and 7 pathways involved in "circulatory system" were enriched in clone A+. Significantly, we revealed the differential expression changes of IFN system genes and immunoglobulin (Ig) genes among the survivors of three clones. Finally, myosins and Igs were identified as co-expression modules which were positively or negatively correlated to CaHV viral loads respectively. CONCLUSIONS Our results revealed the common and distinct gene expression changes in immune and circulatory system in the survivors of three gibel carp gynogenetic clones with different CaHV resistances. The current study represents a paradigm of differential innate and adaptive immune reactions in teleost, and will be beneficial to the disease-resistance breeding of gibel carp.
Collapse
Affiliation(s)
- Wei-Jia Lu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan, 430072 Hubei China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Fan-Xiang Gao
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan, 430072 Hubei China
- University of Chinese Academy of Sciences, Beijing, 100049 China
- Institute of Marine Biology, College of Oceanography, Hohai University, Nanjing, 210098 China
| | - Yang Wang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan, 430072 Hubei China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Qi-Ya Zhang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan, 430072 Hubei China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Zhi Li
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan, 430072 Hubei China
| | - Xiao-Juan Zhang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan, 430072 Hubei China
| | - Li Zhou
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan, 430072 Hubei China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Jian-Fang Gui
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan, 430072 Hubei China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| |
Collapse
|
21
|
Truong D, Boddy KC, Canadien V, Brabant D, Fairn GD, D'Costa VM, Coyaud E, Raught B, Pérez-Sala D, Park WS, Heo WD, Grinstein S, Brumell JH. Salmonella
exploits host Rho GTPase signalling pathways through the phosphatase activity of SopB. Cell Microbiol 2018; 20:e12938. [DOI: 10.1111/cmi.12938] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 06/11/2018] [Accepted: 07/06/2018] [Indexed: 12/20/2022]
Affiliation(s)
- Dorothy Truong
- Cell Biology Program; Hospital for Sick Children; Toronto ON Canada
- Department of Molecular Genetics; University of Toronto; Toronto ON Canada
| | - Kirsten C. Boddy
- Cell Biology Program; Hospital for Sick Children; Toronto ON Canada
- Institute of Medical Science; University of Toronto; Toronto ON Canada
| | | | - Danielle Brabant
- Cell Biology Program; Hospital for Sick Children; Toronto ON Canada
| | - Gregory D. Fairn
- Institute of Medical Science; University of Toronto; Toronto ON Canada
- Keenan Research Centre for Biomedical Science; St. Michael's Hospital; Toronto ON Canada
| | | | - Etienne Coyaud
- Princess Margaret Cancer Centre; University Health Network; Toronto Ontario Canada
| | - Brian Raught
- Princess Margaret Cancer Centre; University Health Network; Toronto Ontario Canada
- Department of Medical Biophysics; University of Toronto; Toronto Ontario Canada
| | - Dolores Pérez-Sala
- Department of Structural and Chemical Biology; Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas; Madrid Spain
| | - Wei Sun Park
- Department of Biological Sciences; Korea Advanced Institute of Science and Technology (KAIST); Daejeon Republic of Korea
| | - Won Do Heo
- Department of Biological Sciences; Korea Advanced Institute of Science and Technology (KAIST); Daejeon Republic of Korea
- Center for Cognition and Sociality; Institute of Basic Science (IBS); Daejeon Republic of Korea
| | - Sergio Grinstein
- Cell Biology Program; Hospital for Sick Children; Toronto ON Canada
- Institute of Medical Science; University of Toronto; Toronto ON Canada
- Keenan Research Centre for Biomedical Science; St. Michael's Hospital; Toronto ON Canada
- Department of Biochemistry; University of Toronto; Toronto ON Canada
| | - John H. Brumell
- Cell Biology Program; Hospital for Sick Children; Toronto ON Canada
- Department of Molecular Genetics; University of Toronto; Toronto ON Canada
- Institute of Medical Science; University of Toronto; Toronto ON Canada
- Sickkids IBD Centre; Hospital for Sick Children; Toronto ON Canada
| |
Collapse
|
22
|
Amado-Azevedo J, de Menezes RX, van Nieuw Amerongen GP, van Hinsbergh VWM, Hordijk PL. A functional siRNA screen identifies RhoGTPase-associated genes involved in thrombin-induced endothelial permeability. PLoS One 2018; 13:e0201231. [PMID: 30048510 PMCID: PMC6062096 DOI: 10.1371/journal.pone.0201231] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 07/11/2018] [Indexed: 12/18/2022] Open
Abstract
Thrombin and other inflammatory mediators may induce vascular permeability through the disruption of adherens junctions between adjacent endothelial cells. If uncontrolled, hyperpermeability leads to an impaired barrier, fluid leakage and edema, which can contribute to multi-organ failure and death. RhoGTPases control cytoskeletal dynamics, adhesion and migration and are known regulators of endothelial integrity. Knowledge of the precise role of each RhoGTPase, and their associated regulatory and effector genes, in endothelial integrity is incomplete. Using a combination of a RNAi screen with electrical impedance measurements, we quantified the effect of individually silencing 270 Rho-associated genes on the barrier function of thrombin-activated, primary endothelial cells. Known and novel RhoGTPase-associated regulators that modulate the response to thrombin were identified (RTKN, TIAM2, MLC1, ARPC1B, SEPT2, SLC9A3R1, RACGAP1, RAPGEF2, RHOD, PREX1, ARHGEF7, PLXNB2, ARHGAP45, SRGAP2, ARHGEF5). In conclusion, with this siRNA screen, we confirmed the roles of known regulators of endothelial integrity but also identified new, potential key players in thrombin-induced endothelial signaling.
Collapse
Affiliation(s)
- Joana Amado-Azevedo
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands
| | - Renee X. de Menezes
- Department of Epidemiology and Biostatistics, VU University Medical Center, Amsterdam, The Netherlands
| | | | - Victor W. M. van Hinsbergh
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands
| | - Peter L. Hordijk
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands
- * E-mail:
| |
Collapse
|
23
|
Abstract
The Rho GTPases were discovered more than 30 years ago, and they were for a long time considered to follow simple cycling between GDP-bound and GTP-bound conformations, as for the Ras subfamily of small GTPases. The Rho GTPases consist of 20 members, but at least 10 of these do not follow this classical GTPase cycle. Thus, based on their kinetic properties, these Rho GTPases can instead be classified as atypical. Some of these atypical Rho GTPases do not hydrolyze GTP, and some have significantly increased intrinsic GDP/GTP exchange activity. This review focuses on this latter category of atypical Rho GTPases, the so-called 'fast-cycling' Rho GTPases. The different members of these fast-cycling atypical Rho GTPases are described in more detail here, along with their potential regulatory mechanisms. Finally, some insights are provided into the involvement of the atypical Rho GTPases in human pathologies.
Collapse
Affiliation(s)
- Pontus Aspenström
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Nobels väg 16 , Box 280, SE, Stockholm, Sweden
| |
Collapse
|
24
|
Harrington EO, Vang A, Braza J, Shil A, Chichger H. Activation of the sweet taste receptor, T1R3, by the artificial sweetener sucralose regulates the pulmonary endothelium. Am J Physiol Lung Cell Mol Physiol 2017; 314:L165-L176. [PMID: 28971978 PMCID: PMC5866431 DOI: 10.1152/ajplung.00490.2016] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
A hallmark of acute respiratory distress syndrome (ARDS) is pulmonary vascular permeability. In these settings, loss of barrier integrity is mediated by cell-contact disassembly and actin remodeling. Studies into molecular mechanisms responsible for improving microvascular barrier function are therefore vital in the development of therapeutic targets for reducing vascular permeability in ARDS. The sweet taste receptor T1R3 is a G protein-coupled receptor, activated following exposure to sweet molecules, to trigger a gustducin-dependent signal cascade. In recent years, extraoral locations for T1R3 have been identified; however, no studies have focused on T1R3 within the vasculature. We hypothesize that activation of T1R3, in the pulmonary vasculature, plays a role in regulating endothelial barrier function in settings of ARDS. Our study demonstrated expression of T1R3 within the pulmonary vasculature, with a drop in expression levels following exposure to barrier-disruptive agents. Exposure of lung microvascular endothelial cells to the intensely sweet molecule sucralose attenuated LPS- and thrombin-induced endothelial barrier dysfunction. Likewise, sucralose exposure attenuated bacteria-induced lung edema formation in vivo. Inhibition of sweet taste signaling, through zinc sulfate, T1R3, or G-protein siRNA, blunted the protective effects of sucralose on the endothelium. Sucralose significantly reduced LPS-induced increased expression or phosphorylation of the key signaling molecules Src, p21-activated kinase (PAK), myosin light chain-2 (MLC2), heat shock protein 27 (HSP27), and p110α phosphatidylinositol 3-kinase (p110αPI3K). Activation of T1R3 by sucralose protects the pulmonary endothelium from edemagenic agent-induced barrier disruption, potentially through abrogation of Src/PAK/p110αPI3K-mediated cell-contact disassembly and Src/MLC2/HSP27-mediated actin remodeling. Identification of sweet taste sensing in the pulmonary vasculature may represent a novel therapeutic target to protect the endothelium in settings of ARDS.
Collapse
Affiliation(s)
- Elizabeth O Harrington
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center , Providence, Rhode Island.,Department of Medicine, Alpert Medical School of Brown University , Providence, Rhode Island
| | - Alexander Vang
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center , Providence, Rhode Island
| | - Julie Braza
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center , Providence, Rhode Island.,Department of Medicine, Alpert Medical School of Brown University , Providence, Rhode Island
| | - Aparna Shil
- Biomedical Research Group, Anglia Ruskin University , Cambridge , United Kingdom
| | - Havovi Chichger
- Biomedical Research Group, Anglia Ruskin University , Cambridge , United Kingdom
| |
Collapse
|
25
|
Bembenek JN, Meshik X, Tsarouhas V. Meeting report - Cellular dynamics: membrane-cytoskeleton interface. J Cell Sci 2017; 130:2775-2779. [PMID: 29360626 DOI: 10.1242/jcs.208660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The first ever 'Cellular Dynamics' meeting on the membrane-cytoskeleton interface took place in Southbridge, MA on May 21-24, 2017 and was co-organized by Michael Way, Elizabeth Chen, Margaret Gardel and Jennifer Lippincott-Schwarz. Investigators from around the world studying a broad range of related topics shared their insights into the function and regulation of the cytoskeleton and membrane compartments. This provided great opportunities to learn about key questions in various cellular processes, from the basic organization and operation of the cell to higher-order interactions in adhesion, migration, metastasis, division and immune cell interactions in different model organisms. This unique and diverse mix of research interests created a stimulating and educational meeting that will hopefully continue to be a successful meeting for years to come.
Collapse
Affiliation(s)
- Joshua N Bembenek
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, TN 37996, USA
| | - Xenia Meshik
- Department of Anesthesiology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Vasilios Tsarouhas
- Department of Molecular Biosciences, Stockholm University, Stockholm, Sweden
| |
Collapse
|